Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nephron ; 135(4): 291-306, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28064277

RESUMEN

BACKGROUND: The role of transforming growth factor-ß (TGF-ß) has recently gained much attention in diabetic nephropathy and kidney fibrosis. In this study, we extend this to an assessment of transcriptional regulation of the entire TGF-ß superfamily in kidneys from diabetic vs. healthy mice. In order to study the translation between mouse model and patients, we evaluated the signature of phosphorylated Sma- and Mad-related protein 2 (pSmad2), as molecular marker of TGF-ß/activin activity, in the kidneys of streptozotocin (STZ)-treated mice compared to that of type 1 diabetes (T1D) patients. METHODS: Patterns of pSmad2 were determined in kidneys from T1D patients with progressed diabetic nephropathy (DN), defined by hyperglycemia, microalbuminuria, and increased levels of serum creatinine. They were compared to changes seen in the STZ-induced DN mouse model. This was studied by immunohistochemistry (IHC) with an antibody specific for pSmad2. Diabetic mice were also characterized by pSmad1/5/8 (IHC), pSmad2/3 (flow cytometry), and TGF-ß family members including bone morphogenetic protein (BMP)-like proteins (quantitative real-time polymerase chain reaction [qPCR]). RESULTS: Renal tubules in DN patients and in STZ mice showed upregulation of pSmad2 concomitant with significantly enlarged distal tubule lumens (p < 0.0001). Renal-derived CD11b+ cells from STZ mice showed elevated pSmad2/3, while endothelial cells had reduced pSmad2/3 levels. No pSmad1/5/8 was observed in the tubule compartment of STZ-treated mice. On total kidney mRNA level, a signature favoring activation of the TGF-ß/activin pathway and inhibition of the BMP pathway was demonstrated by qPCR. CONCLUSION: Although the pre-clinical DN model lacks the features of fibrosis present in human DN, both species show induction of a local milieu favoring pSmad2 signaling, which may be useful as a disease biomarker in pre-clinical models.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Activinas/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Proteínas Morfogenéticas Óseas/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Masculino , Ratones , Ratones de la Cepa 129 , Persona de Mediana Edad , Modelos Biológicos , Fosforilación , Proteína Smad2/sangre , Proteína smad3/sangre , Factor de Crecimiento Transformador beta/genética , Regulación hacia Arriba
2.
Biochem Soc Trans ; 44(4): 1142-9, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27528762

RESUMEN

Angiogenesis is a hallmark of cancer and is now a validated therapeutic target in the clinical setting. Despite the initial success, anti-angiogenic compounds impinging on the vascular endothelial growth factor (VEGF) pathway display limited survival benefits in patients and resistance often develops due to activation of alternative pathways. Thus, finding and validating new targets is highly warranted. Activin receptor-like kinase (ALK)1 is a transforming growth factor beta (TGF-ß) type I receptor predominantly expressed in actively proliferating endothelial cells (ECs). ALK1 has been shown to play a pivotal role in regulating angiogenesis by binding to bone morphogenetic protein (BMP)9 and 10. Two main pharmacological inhibitors, an ALK1-Fc fusion protein (Dalantercept/ACE-041) and a fully human antibody against the extracellular domain of ALK1 (PF-03446962) are currently under clinical development. Herein, we briefly recapitulate the role of ALK1 in blood vessel formation and the current status of the preclinical and clinical studies on inhibition of ALK1 signalling as an anti-angiogenic strategy. Future directions in terms of new combination regimens will also be presented.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Neoplasias/irrigación sanguínea , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal/efectos de los fármacos
3.
Clin Cancer Res ; 22(1): 96-106, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26373572

RESUMEN

PURPOSE: Antiangiogenic therapy, mostly targeting VEGF, has been applied in cancer patients for the last decade. However, resistance to anti-VEGF therapy and/or no significant benefit as monotherapeutic agent is often observed. Therefore, new antiangiogenic strategies are needed. In the current study, we investigated the therapeutic effect of interfering with the bone morphogenetic protein (BMP)9/activin receptor-like kinase (ALK)1 signaling pathway by using an ALK1-Fc ligand trap. EXPERIMENTAL DESIGN: We analyzed the potential antiangiogenic and antitumor effects of ALK1-Fc protein as monotherapy and in combination with chemotherapy in vivo in mouse models of melanoma, head and neck cancer, and invasive lobular breast carcinomas. ALK1-Fc sequesters BMP9 and 10 and prevents binding of these ligands to endothelial ALK1, which regulates angiogenesis. RESULTS: Treatment of mice with ALK1-Fc strongly decreased the tumors' microvascular density in the three different mouse cancer models. However, this effect was not accompanied by a reduction in tumor volume. An immunohistochemical analysis of the tumor samples revealed that ALK1-Fc treatment increased the pericyte coverage of the remaining tumor vessels and decreased the hypoxia within the tumor. Next, we observed that combining ALK1-Fc with cisplatin inhibited tumor growth in the breast and head and neck cancer models more efficiently than chemotherapy alone. CONCLUSIONS: The addition of ALK1-Fc to the cisplatin treatment was able to enhance the cytotoxic effect of the chemotherapy. Our results provide strong rationale to explore combined targeting of ALK1 with chemotherapy in a clinical setting, especially in the ongoing phase II clinical trials with ALK1-Fc.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/farmacología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Factor 2 de Diferenciación de Crecimiento/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/farmacología , Ratones , Ratones Noqueados , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neovascularización Patológica/genética , Proteínas Recombinantes de Fusión/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Carga Tumoral
4.
Arterioscler Thromb Vasc Biol ; 35(3): 616-27, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25633317

RESUMEN

OBJECTIVE: Arterial calcification is considered a major cause of death and disabilities worldwide because the associated vascular remodeling leads to myocardial infarction, stroke, aneurysm, and pulmonary embolism. This process occurs via poorly understood mechanisms involving a variety of cell types, intracellular mediators, and extracellular cues within the vascular wall. An inverse correlation between endothelial primary cilia and vascular calcified areas has been described although the signaling mechanisms involved remain unknown. We aim to investigate the signaling pathways regulated by the primary cilium that modulate the contribution of endothelial cells to vascular calcification. APPROACH AND RESULTS: We found that human and murine endothelial cells lacking primary cilia are prone to undergo mineralization in response to bone morphogenetic proteins stimulation in vitro. Using the Tg737(orpk/orpk) cillium-defective mouse model, we show that nonciliated aortic endothelial cells acquire the ability to transdifferentiate into mineralizing osteogenic cells, in a bone morphogenetic protein-dependent manner. We identify ß-CATENIN-induced SLUG as a key transcription factor controlling this process. Moreover, we show that the endothelial expression of SLUG is restricted to atheroprone areas in the aorta of LDLR(-/-) mice. Finally, we demonstrate that SLUG and phospho-homolog of the Drosophila protein, mothers against decapentaplegic (MAD) and the Caenorhabditis elegans protein SMA (from gene sma for small body size)-1/5/8 expression increases in endothelial cells constituting the vasa vasorum in the human aorta during the progression toward atherosclerosis. CONCLUSIONS: We demonstrated that the lack of primary cilia sensitizes the endothelium to undergo bone morphogenetic protein-dependent-osteogenic differentiation. These data emphasize the role of the endothelial cells on the vascular calcification and uncovers SLUG as a key target in atherosclerosis.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Células Endoteliales/metabolismo , Factores de Transcripción/metabolismo , Calcificación Vascular/metabolismo , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proteínas Morfogenéticas Óseas/metabolismo , Transdiferenciación Celular , Células Cultivadas , Cilios , Modelos Animales de Enfermedad , Células Endoteliales/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones Noqueados , Ratones Mutantes , Mutación , Osteoblastos/metabolismo , Osteogénesis , Fosforilación , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal , Proteínas Smad Reguladas por Receptores/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Transfección , Proteínas Supresoras de Tumor/genética , Calcificación Vascular/genética , Calcificación Vascular/patología , beta Catenina/metabolismo
5.
Nat Commun ; 5: 3388, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24584437

RESUMEN

In advanced cancers, the TGF-ß pathway acts as an oncogenic factor and is considered to be a therapeutic target. Here using a genome-wide cDNA screen, we identify nuclear receptor NR4A1 as a strong activator of TGF-ß signalling. NR4A1 promotes TGF-ß/SMAD signalling by facilitating AXIN2-RNF12/ARKADIA-induced SMAD7 degradation. NR4A1 interacts with SMAD7 and AXIN2, and potently and directly induces AXIN2 expression. Whereas loss of NR4A1 inhibits TGF-ß-induced epithelial-to-mesenchymal transition and metastasis, slight NR4A1 ectopic expression stimulates metastasis in a TGF-ß-dependent manner. Importantly, inflammatory cytokines potently induce NR4A1 expression, and potentiate TGF-ß-mediated breast cancer cell migration, invasion and metastasis in vitro and in vivo. Notably, NR4A1 expression is elevated in breast cancer patients with high immune infiltration and its expression weakly correlates with phosphorylated SMAD2 levels, and is an indicator of poor prognosis. Our results uncover inflammation-induced NR4A1 as an important determinant for hyperactivation of pro-oncogenic TGF-ß signalling in breast cancer.


Asunto(s)
Neoplasias Mamarias Animales/metabolismo , Metástasis de la Neoplasia/fisiopatología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Femenino , Inmunohistoquímica , Neoplasias Mamarias Animales/genética , Ratones , Ratones Noqueados , Metástasis de la Neoplasia/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/genética , Ubiquitinación/genética , Ubiquitinación/fisiología , Pez Cebra
6.
Curr HIV Res ; 8(6): 418-29, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20636281

RESUMEN

The persistence of latent HIV-infected cellular reservoirs represents the major hurdle to virus eradication on patients treated with HAART. It has been suggested that successful depletion of such latent reservoirs will require a combination of therapeutic agents that can specifically and efficiently act on cells harboring latent HIV-1 provirus. Using Jurkat-LAT-GFP cells, a tractable model of HIV-1 latency, we have found that bryostatin -1 reactivates HIV-1 through a classical PKC-dependent pathway. Bryostatin-1 also activates MAPKs and NF-κB pathways and synergizes with HDAC inhibitors to reactivate HIV-1 from latency. Bryostatin-1 downregulates the expression of the HIV-1 co-receptors CD4 and CXCR4 and prevented de novo HIV-1 infection in susceptible cells. We applied proteomic methods to investigate major changes in protein expression in Jurkat-LAT-GFP under latency and reactivation conditions. We identified up-regulation of proteins that may be involved in the innate anti-HIV-1 response (NKEF-A and MHD2) and in different cell functions (i.e. cofilin-1 and transgelin-2) of the host cells. PKC agonists may represent a valuable pharmacological approach to purge latent HIV from cellular reservoirs and at the moment, the only clinically available PKC agonist is bryostatin-1. This drug has been tested in numerous clinical trials and its pharmacokinetics and toxicity in humans is well known. Moreover, bryostatin-1 potently synergizes with other HDAC inhibitors commonly used in the medical practice such as valproic acid. Therefore, bryostatin-1, alone or in combination with HDAC inhibitors, could be used in HAART treated patients to validate the hypothesis that reactivating HIV-1 from latency could purge HIV-1 reservoirs.


Asunto(s)
Brioestatinas/farmacología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/fisiología , Provirus/fisiología , Latencia del Virus/efectos de los fármacos , Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/uso terapéutico , Terapia Antirretroviral Altamente Activa , Antígenos CD4/metabolismo , Regulación hacia Abajo , Infecciones por VIH/inmunología , Inhibidores de Histona Desacetilasas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Células Jurkat , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Proteína Quinasa C/metabolismo , Provirus/efectos de los fármacos , Provirus/metabolismo , Receptores CXCR4/metabolismo , Regulación hacia Arriba
7.
Biochem Pharmacol ; 79(12): 1805-14, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20206142

RESUMEN

Cerebral microvascular endothelial cells play an active role in maintaining cerebral blood flow, microvascular tone and blood brain barrier (BBB) functions. Endogenous N-acyl-dopamines like N-arachidonoyl-dopamine (NADA) and N-oleoyl-dopamine (OLDA) have been recently identified as a new class of brain neurotransmitters sharing endocannabinoid and endovanilloid biological activities. Endocannabinoids are released in response to pathogenic insults and may play an important role in neuroprotection. In this study we demonstrate that NADA differentially regulates the release of PGE(2) and PGD(2) in the microvascular brain endothelial cell line, b.end5. We found that NADA activates a redox-sensitive p38 MAPK pathway that stabilizes COX-2 mRNA resulting in the accumulation of the COX-2 protein, which depends on the dopamine moiety of the molecule and that is independent of CB(1) and TRPV1 activation. In addition, NADA inhibits the expression of mPGES-1 and the release of PGE(2) and upregulates the expression of L-PGD synthase enhancing PGD(2) release. Hence, NADA and other molecules of the same family might be included in the group of lipid mediators that could prevent the BBB injury under inflammatory conditions and our findings provide new mechanistic insights into the anti-inflammatory activities of NADA in the central nervous system and its potential to design novel therapeutic strategies to manage neuroinflammatory diseases.


Asunto(s)
Ácidos Araquidónicos/farmacología , Encéfalo/metabolismo , Ciclooxigenasa 2/metabolismo , Dopamina/análogos & derivados , Células Endoteliales/efectos de los fármacos , ARN Mensajero/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Encéfalo/citología , Línea Celular , Ciclooxigenasa 2/genética , Dopamina/farmacología , Células Endoteliales/enzimología , Inducción Enzimática/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , ARN Mensajero/genética
8.
EMBO J ; 28(13): 1843-54, 2009 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-19521338

RESUMEN

Wnt signalling is a crucial signalling pathway controlling intestinal homeostasis and cancer. We show here that the JNK MAP kinase pathway and one of its most important substrates, the AP-1 transcription factor c-Jun, modulates Wnt signalling strength in the intestine. Transgenic gut-specific augmentation of JNK signalling stimulated progenitor cell proliferation and migration, resulting in increased villus length. In the crypt, c-Jun protein was highly expressed in progenitor cells and the absence of c-Jun resulted in decreased proliferation and villus length. In addition to several known c-Jun/AP-1 target genes, expression of Wnt target genes Axin2 and Lgr5 were stimulated by JNK activation, suggesting a cross talk of JNK to Wnt signalling. Expression of the Wnt pathway component TCF4 was controlled by JNK activity, and chromatin immunoprecipitation and reporter assays identified tcf4 as a direct c-Jun target gene. Consequently, increased JNK activity accelerated tumourigenesis in a model of colorectal carcinogenesis. As c-jun is a direct target of the TCF4/beta-catenin complex, the control of tcf4 expression by JNK/c-Jun leads to a positive feedback loop that connects JNK and Wnt signalling. This mechanism regulates the physiological function of progenitor cells and oncogenic transformation.


Asunto(s)
Carcinoma/fisiopatología , Neoplasias del Colon/fisiopatología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Transcripción AP-1/metabolismo , Proteínas Wnt/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Intestinos/citología , Intestinos/fisiopatología , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas c-jun/genética , Transducción de Señal , Células Madre/citología , Factores de Transcripción TCF/genética , Factor de Transcripción 4 , Factor de Transcripción AP-1/genética , Proteínas Wnt/genética
9.
Biochem Pharmacol ; 77(8): 1401-9, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19426679

RESUMEN

Denbinobin, a 1,4-phenanthrenequinone firstly isolated from the stems of Dendrobium moniliforme (Shi-Hu in Chinese medicine), has been reported to exhibit anti-tumoral and anti-inflammatory activities through mechanism(s) not yet fully understood. Because of the critical role of the transcription factor NF-kappaB and of ROS-induced activation of stress regulated kinases in tumorigenesis, we have investigated the effect of denbinobin on these pathways. We found that denbinobin is a potent inhibitor of TNFalpha and PMA-induced NF-kappaB activation, and that it can block the phosphorylation and degradation of IkappaBalpha by inhibiting TAK1 activity, an event lying upstream of IKK activation. Moreover, treatment with denbinobin not only elicited apoptotic signalling, including mitochondrial membrane dysfunction, activation of caspases and cleavage of poly(ADP-ribose) polymerase, but also induced intracellular reactive oxygen species (ROS) generation and sustained activation of the mitogen-activated kinases (MAPKs) ERK1+2, p38 and JNK 1+2. The apoptotic effects of denbinobin could be prevented by pre-treatment with the intracellular ROS scavenger N-acetyl-l-cysteine, but not by pharmacological inhibition of MAPKs, suggesting that intracellular ROS generation underlies denbinobin-induced apoptosis, and that this effect takes place in an MAPKs-independent pathway. To define the structural elements critical for these activities, a series of phenanthrenequinones with different substituents in the phenanthrene- and/or in the quinone ring were prepared and assayed for NF-kappaB inhibition and ROS production. In this way, the major structure-activity relationships and the structural elements critical for the activity of denbinobin could be established.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Fenantrenos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Antraquinonas/síntesis química , Antraquinonas/aislamiento & purificación , Antineoplásicos Fitogénicos/síntesis química , Antineoplásicos Fitogénicos/aislamiento & purificación , Western Blotting , Cannabis/química , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dendrobium/química , Ensayo de Cambio de Movilidad Electroforética , Humanos , Células Jurkat , Potencial de la Membrana Mitocondrial/efectos de los fármacos , FN-kappa B/biosíntesis , Fenantrenos/síntesis química , Fenantrenos/aislamiento & purificación , Tallos de la Planta/química , Plásmidos , Transfección
10.
J Neurochem ; 109(2): 452-64, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19200337

RESUMEN

It is widely accepted that neuroinflammation is a key player in various pathological events associated with brain injury. More specifically, glial activation and the subsequent release of pro-inflammatory cytokines, reactive oxygen species (ROS), and prostaglandins play a role of paramount importance in cerebral damage. In this study, we examined the role of two endocannabinoids, anandamide (AEA) and N-arachidonoyldopamine (NADA) in the regulation of prostaglandin E(2) (PGE(2)) synthesis in primary glial cells. We show that NADA is a potent inhibitor of PGE(2) synthesis in lipopolysaccharide (LPS) stimulated cells, without modifying the expression or enzymatic activity of COX-2 and the production of prostaglandin D(2). We also show that NADA has the ability to prevent the free radical formation in primary microglial cells. The key findings of this investigation are our observation that AEA and NADA have opposite effects on glial cells and, most importantly, the first description of NADA as a potential antioxidative and anti-inflammatory agent acting through a mechanism that involves reduction in the synthesis of microsomal prostaglandin E synthase in LPS-activated microglia. These findings provide new mechanistic insights into the anti-inflammatory activities of NADA in the CNS and its potential to design novel therapeutic strategies to manage neuroinflammatory diseases.


Asunto(s)
Ácidos Araquidónicos/fisiología , Dinoprostona/análogos & derivados , Dinoprostona/biosíntesis , Dopamina/análogos & derivados , Isoprostanos/biosíntesis , Neuroglía/metabolismo , Animales , Animales Recién Nacidos , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Antioxidantes/química , Antioxidantes/metabolismo , Antioxidantes/fisiología , Ácidos Araquidónicos/química , Ácidos Araquidónicos/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Dinoprostona/química , Dinoprostona/metabolismo , Dopamina/química , Dopamina/metabolismo , Dopamina/fisiología , Endocannabinoides , Isomerismo , Isoprostanos/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroglía/química , Neuroglía/efectos de los fármacos , Alcamidas Poliinsaturadas/química , Alcamidas Poliinsaturadas/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Biochem Pharmacol ; 76(10): 1240-50, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18840408

RESUMEN

Anthraquinones and structurally related compounds have been recently shown to exert antiviral activities and thus exhibit a therapeutic potential. In this study we report the isolation of the 1,4-phenanthrenequinone, denbinobin, from a variety of Cannabis sativa. Denbinobin does not affect the reverse transcription and integration steps of the viral cycle but prevents HIV-1 reactivation in Jurkat T cells activated by TNFalpha, mAbs anti-CD3/CD28 or PMA. In addition, denbinobin inhibits HIV-1-LTR activity at the level of transcription elongation and also TNFalpha-induced HIV-1-LTR transcriptional activity. We found that denbinobin prevents the binding of NF-kappaB to DNA and the phosphorylation and degradation of NF-kappaB inhibitory protein, IkappaBalpha, and inhibits the phosphorylation of the NF-kappaB p65 subunit in TNFalpha-stimulated cells. These results highlight the potential of the NF-kappaB transcription factor as a target for natural anti-HIV-1 compounds such as 1,4-phenanthrenequinones, which could serve as lead compounds for the development of an alternative therapeutic approach against AIDS.


Asunto(s)
Antraquinonas/farmacología , VIH-1/efectos de los fármacos , FN-kappa B/metabolismo , Fenantrenos/farmacología , Replicación Viral/efectos de los fármacos , Antraquinonas/química , Antraquinonas/aislamiento & purificación , VIH-1/fisiología , Humanos , Células Jurkat , FN-kappa B/antagonistas & inhibidores , Fenantrenos/química , Fenantrenos/aislamiento & purificación , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Replicación Viral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...